Open Access

Exosomes as carriers transporting long non‑coding RNAs: Molecular characteristics and their function in cancer (Review)

  • Authors:
    • Damian Kołat
    • Raneem Hammouz
    • Andrzej K. Bednarek
    • Elżbieta Płuciennik
  • View Affiliations

  • Published online on: June 5, 2019     https://doi.org/10.3892/mmr.2019.10340
  • Pages: 851-862
  • Copyright : © Kołat et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Long non‑coding RNAs (lncRNAs) comprise a sizeable class of non‑coding RNAs with a length of over 200 base pairs. Little is known about their biological function, although over 20,000 lncRNAs have been annotated in the human genome. Through a diverse range of mechanisms, their primary function is in the regulation of the transcription of protein‑coding genes. lncRNA transcriptional activation can result from a group of nucleus‑retained and chromatin‑associated lncRNAs, which function as scaffolds in the cis/trans recruitment of transcription factors, co‑activators or chromatin remodelers, and/or promoter enhancers. Exosomes are released as extracellular vesicles and they are produced by endocytic pathways. Their synthesis is initiated by various processes including ceramide synthesis, release of intracellular Ca2+ or acid‑base balance disorders. Prior to vesicle creation, selective cargo loading occurs in the Endosomal Sorting Complex Required for Transport. Participation of endosomal sorting proteins such as tetraspanins or specific sumoylated proteins required for transport has been indicated in research. The endosomal‑sorting complex consists of four components, these induce the formation of multivesicular bodies and the induction of membrane deformation to form exosomes. Nanovesicles could be formed inside multivesicular bodies to allow transport outside the cell or digestion in lysosomes. The molecular content of exosomes is more heterogenic than its synthesis process, with different cargoes being examined inside vesicles with regard to the type or stage of cancers. This paper will review the importance of lncRNAs as crucial molecular content of exosomes, indicating its involvement in tumour suppression, pro‑tumorigenic events and the development of novel therapeutic approaches in the near future. Further studies of their mechanisms of function are essential, as well as overcoming several challenges to gain a clearer insight to the approaches for the best clinical application.

Introduction

The term transcriptome encompasses the complete set of RNA molecules applying to an entire organism or specific cell type, making it more dynamic than the genome. It comprises several types of transcripts which are responsible for changes in gene expression. Two major classes of regulatory non-coding RNAs (ncRNAs) which lack any transcriptive capability include microRNAs and long non-coding RNASs (lncRNAs). They play a significant role in diverse cellular processes from enabling normal development to disease progression, as well as cell communication. A valuable example of a biological carrier that can transport abundant quantities of lncRNA is that of the exosome: A type of extracellular vesicle involved in cell-to-cell communication and disease transmission. Its role has been documented in both physiological conditions and pathological changes such as cancer development, where they are responsible for the regulation of various processes including immunosuppression, proliferation and induction of pre-metastatic niche formation. In this review, the role of a number of exosomal lncRNA molecules will be discussed.

Characteristics of exosomes

The first scientific report to examine the possibility that intracellular structures could be associated with the process of budding vesicles bearing receptors during maturation, using ferritin-labelled proteins and anti-IgG, recorded the externalization of transferrin receptors into the intercellular space during sheep reticulocyte maturation (1). The term exosome was later proposed in 1987 (2).

Exosomes are small spherical nanovesicles ranging from 20 to 120 nm (3,4). They belong to the group of membrane-derived vesicles known as extracellular vesicles (EVs), together with oncosomes, ectosomes or apoptotic bodies (5). The classification of each vesicle to its specific group depends on genesis, composition, size and density (see Table I). Extracellular vesicles can be secreted constitutively or as a result of specific induction (6). The origin of exosomes is linked to the creation of multivesicular bodies (MVBs) in the endosomal pathway; MVBs are structures with a lipid bilayer and a density of 1.12–1.19 g/Ml (7) and which display 5′ nucleotidase activity (8).

Table I.

Subdivision of extracellular vesicles based on their physiological features [based on (5,14,75)].

Table I.

Subdivision of extracellular vesicles based on their physiological features [based on (5,14,75)].

Extracellular vesiclesSizeDensityOrigin of vesicles
Exosomes20–120 nm1.12–1.19 g/mlMultivesicular body
Ectosomes~0.1–1 µm~1.16 g/mlCell membrane
Apoptotic bodies0.05–5 µm1.24–1.28 g/mlCell membrane
Oncosomes1–10 µmNot specifiedCell membrane

Exosomes are released from many types of healthy or tumour cells, although more abundantly in the latter (9), and can be found in the tumour microenvironment (TME) and blood samples of cancer patients (10). Physiologically, exosomes can be found in urine (10), sperm (3), amniotic fluid (9) and various other biological fluids. The role of exosomes in metastasis, angiogenesis and epithelial-to-mesenchymal transition (EMT) has been confirmed in cancer development by various studies (9). Exosomes released by tumour cells were shown to constitute a leading source of antigens for dendritic cells inducing antitumour immune responses (11). The physiological functions of exosomes include cellular communication, immunity, infections, elimination of waste products and signalling (12). There is also evidence of their participation in aging-related diseases i.e. neurodegeneration when different types of mRNAs, miRNAs or lncRNA change their abundance inside nanovesicles (13).

Biogenesis

Exosome secretions and their molecular mechanisms are rather obscure, since their release is regulated by several signal molecules and various processes including ceramide synthesis and Ca2+signalling (3). Proteins that interact with the cell membrane and perform lipid bilayer deformation are epsin, amphiphysin, endofilin, syndapin and dynamin (4). The biogenesis follows the following sequence: The process begins with membrane invagination and exosome release to cytoplasm, followed by early endosome (EE) formation, and then the formation of intraluminal vesicles (ILVs) inside the late endosome (LE).

Under both normal and pathological conditions, exosomes have an endocytic origin and are released by different types of cells. Although exosomes typically mimic the composition of the parent cells, they may contain a few signature proteins. Following the formation of exosomes within the endocytic pathway, they are released from the plasma membrane via MVBs from early endosome maturation and may further fuse with lysosomes or the plasma membrane. The most common and well-known mechanism of cargo sorting is based on the action of proteins from Endosomal Sorting Complex Required for Transport (ESCRT) (6). When this pathway is inactivated, there is a possibility to recruit sumoylated protein hnRNPA2B1 [it binds microRNA (14)] or maintain the signal through tetraspanin-enriched microdomains (TEMs) composed of transmembrane glycoproteins (8,15). The schematic of ESCRT complex based on yeast, is shown in Fig. 1.

The most important proteins associated with ESCRT are: ALIX (ALG-2-interacting protein X), syndecan and syntenin (16). Syntenin ends the formation process through syndecan sorting and binding to ALIX, which affects ESCRT-III (4). Direct ALIX-syntenin interaction is possible through LYPX(n)L motifs (16), which are observed in the viral proteins found in late assembly domains (L-domains), these being responsible for recruiting the host ESCRT machinery directly or via ESCRT-associated proteins. The final product of this ESCRT and auxiliary protein activity is the creation of an MVB containing exosomes.

Despite general syntenin-syndecan-ALIX axis, there are some other molecular individuals involved in controlling ESCRT. Examples are ADP-ribosylation factor 6 (ARF6) GTPase and Phospholipase D2 (PLD2) enzyme which are considered as proteins associated with syntenin (17) and are responsible for the control of the budding of ILVs into MVBs (18). LIP5 (LYST-interacting protein 5), also known as vesicle trafficking 1 protein (VTA1), is a cofactor for Vps4 ATPase that enhances Vps4-mediated disassembly of ESCRT-III polymer (19). Cep55 (centrosomal protein 55) can mediate ALIX and ESCRT-I complex recruitment; however, interactions with the components of ESCRT-II and ESCRT-III have also been observed (20). Further examples consist of CD2AP (CD2-associated protein), IQGAP1 (IQ-motif-containing GTPase-activating protein 1) or ROCK1 (Rho-associated kinase 1) which are thought to interact with ALIX or TSG-101 (Tumor susceptibility gene 101). They participate in processes such as cytokinesis (20), viral budding (21) and cell polarity (22), respectively. Finally, enzyme heparanase, able to internally cleave heparan sulfate chains on syndecans, acts by increasing exosome production by stimulating intraluminal budding of syndecan and syntenin (23).

After the ‘mature’ multivesicular body is formed, it can be transported to lysosomes (degraded hydrolytic enzymes) or interact with the lipid membrane, resulting in exosome release outside the cell. In the latter case, the attachment of MVB to the lipid bilayer takes place with the help of Rab-GTP and SNARE family proteins. Examples of these groups are given in Table II.

Table II.

Selected proteins involved in fusion of MVB with cell membrane and in exosome secretion [based on (3,4,7,87)].

Table II.

Selected proteins involved in fusion of MVB with cell membrane and in exosome secretion [based on (3,4,7,87)].

ProteinProteins familyFunction
Rab7; Rab11RabGTPPromotes proper protein attachment in pathways dependent on calcium ions
Rab27 (a, b)RabGTPRegulates the stages of exosome secretion, controls the location of MVB
Rab35RabGTPRegulates exosomes secretion by interacting with GTP-activating protein- TBC1(Tre-2/Bub2/Cdc16)
RAL-1RasGTPRecruiting the Syx-5 protein, thereby mediating in fusion of membranes
Syx-5 (Syntaxin5)SNAREStimulates fusion of MVB with cell membrane
VAMP7SNARENecessary for MVB fusion with cell membrane (Vesicle-Associated Membrane Protein)
YKT6SNAREMediates the release of exosomes from the cell

Following the fusion of MVB and cell membrane, the exosomes can be transported to distant areas of the organism (via blood vessels) or interact with neighbouring cells (4). ‘Targeted’ cells can internalize nanovesicles in endocytic way where the absorption itself is facilitated through phagocytosis, fusion with cell membrane or ligand-receptor interaction. Fig. 2 presents the process of biogenesis, secretion and absorption of exosomes by means of ESCRT machinery.

Molecular content

Nanovesicle formation exhibits some characteristic stages of biogenesis and involves specific structures present in the cell. The selective mechanism of transporting cargo to the exosome interior is a more heterogenous process, as evidenced by their varied content, even in different types and stages of cancer development (8). The composition of exosomes contains countless types of proteins, including cytokines and growth factors, lipids or nucleic acids (3), including characteristic biomarkers such as CD63, CD81, CD9, TSG-101, ALIX or HSP70 which allow direct identification of exosomes (6).

Proteins

The profile of the proteins within the vesicles consists of cytoplasmic proteins, cell membrane proteins or proteins from endosomes; however, no proteins from organelles have been detected (6). These proteins include a number of exosomal molecules, such as those connected with MVB biogenesis (e.g., ALIX, TSG-101) (6), ESCRT components (3), transport and fusion proteins (annexin, Rab-GTP, flotillin) and heat shock cognate (HSC). Other important ones include integrins, tetraspanins or subunits of G proteins. The tumour exosomes (TEXs) include additional molecules that participate in the EMT process including matrix metalloproteinases (MMPs), IL-6, TGF-β and immunosupression factors (FasL, TRAIL and GAL-9 that stimulates T cell apoptosis) (8). Some of them are presented in Table III.

Table III.

Examples of exosomal molecules involved in tumour progression, immunosuppression and apoptosis [based on (3,4,6,10,88,89)].

Table III.

Examples of exosomal molecules involved in tumour progression, immunosuppression and apoptosis [based on (3,4,6,10,88,89)].

Type of molecule in exosomeFunctionType of cells
ITG α6β4; ITG α6β1Connected with metastasis to the lungs, promotes TEXs adhesion in the lungsLung cancer cells
ITG αVβ5Connected with metastasis to the liver, binding to Kupffer cellsLiver cancer cells
TGF-β; IL-10; MCP-1 (Monocyte Chemoattractant Protein-1)Promote cell migrationLung cancer cells, melanoma cells
MHC-II (Major Histocompatibility Complex-II)Stimulation of CD4+ cellsB lymphocytes, DC cells
OVA (Ovalbumin) EGFR (Epidermal Growth Factor Receptor);Inhibition of immune responseMelanoma cells
KRAS (Kirsten RAt Sarcoma viral oncogene homolog)Proliferation, resistance to treatmentLung cancer cells
EGFRvIII (EGFR variant III)Anti-apoptotic abilitiesGlioma cells
MDR-1 (Multi-Drug Resistance)Resistance for drugProstate cancer cells
HER2 (Human Epidermal growth factor Receptor 2)Resistance to treatment with TrastuzumabBreast cancer cells
Lipids

The lipid composition of exosomes is distinct from that of the cell membrane: it is rich in components such as cholesterol, sphingomyelin, glycerophospholipids, phosphatydylinositol, ceramide or phosphatidilserine, as well as prostaglandins and leukotrienes (6). The lipid content in exosomes is higher than that of the parent cell except phosphatidylcholine which is ordinarily decreased (24). An example of exosomal lipid activity is the differentiation of bone-marrow myeloid cells into myeloid-derived suppressor cells (MDSCs) by prostaglandin E2. The accumulation of MDSCs leads to immunosuppression, which provides favourable conditions for metastasis (8). The participation of lipids in the biogenesis of exosomes is summarized below (Table IV).

Table IV.

The examples of lipids involved in formation and secretion of exosomes [based on (9)].

Table IV.

The examples of lipids involved in formation and secretion of exosomes [based on (9)].

LipidFunction
PhosphatidylserineParticipation in microautophagy connected with HSC70 protein (process of exosomes synthesis)
LysoBisPhosphAtidic (LBPA)Change in membrane dynamics (in collaboration with ALIX)
BisMonoacylglycerolPhosphate (BMP)Binding to ALIX protein; (BMP formed by PhosphoLipase D2)
CeramideInvolved in the biosynthesis of exosomes and packaging miRNA into them (in a process independent of the ESCRT machinery)
CholesterolParticipation in secretion of FLOT2-positive exosomes
Lipid rafts (cholesterol-rich sphingolipid microdomains)Participation in the biogenesis of exosomes inside MSCs (Mesenchymal Stem Cells)
Phosphatidylcholine transporter ABCA3Participation in the biogenesis of exosomes derived from B-cell lymphoma
Nucleic acids

Exosomes also can include different types of RNA or DNA such as mRNA, miRNA (6), lncRNA, dsDNA, RNA-ROR (regulator of reprogramming) (3), and therefore play a vital role in regulating signaling pathways and gene expression (refer to Table V). These DNA and RNA molecules may be transferred into recipient cell by the fusion of exosome and cell membranes, aiding the regulation of large numbers of signaling pathways or expression of genes (3). Many studies indicate elevated levels of lncRNA in extracellular exosomes compared to parent cells (25,26). This can be attributed to the action of nucleotide patterns that are present in transcripts as cis-acting elements responsible for targeting them into exosomes (27).

Table V.

The examples of nucleic acids that occur in exosomes [based on (3,6,90)].

Table V.

The examples of nucleic acids that occur in exosomes [based on (3,6,90)].

Nucleic acidLength in base pairs (bp)Function
mRNA~3,250 bp (for TMPRSS2 in prostate cancer, TransMembrane PRotease Serine 2)Participation in metastasis, progression of cancer
lncRNAMore than 200 bpRegulation of gene expression at transcriptional, posttranscriptional and epigenetic levels
dsDNAMore than 2,500 bpParticipation in neoplasm (more frequent occurrences in tumour exosomes than exosomes from normal cells)
RNA-ROR~2,600 bpResistance to chemotherapy in HepatoCellular Carcinoma, promoting cancerogenesis through specific histone methylation

The purpose of this review is to highlight the significance of lncRNA as a pivotal molecule inside exosomes. Therefore, it will summarize the role of various exosomal lncRNAs, together with their function in carcinogenesis and in other biological processes.

lncRNA

The functional annotation of 60,770 full length longer cDNAs and rare transcripts in the mouse transcriptome resulted in the discovery of a novel class of non-coding cDNAs, among which ~80% of the transcripts are not spliced (28). The lncRNAs constitute a class of transcripts that does not encode proteins and whose length exceeds 200 bp (29). This group includes several heterogenous molecules that are difficult to categorize: LncRNAs can be divided into long intergenic non-coding RNAs (lincRNAs), long intronic non-coding RNAs and half-STAU1-binding site RNAs (1/2sbsRNAs) (30). Depending on the DNA coding strand, the lncRNAs can be classified as sense or antisense transcripts (31).

Some resemblance can be found between mRNAs and lncRNAs in some biochemical processes, for example mechanisms of polyadenylation and splicing or participation of RNA polymerase II in the process of their transcription (32). The roles of these molecules are connected with transduction signal, decoys, guides or scaffolds (31); however, their main function is associated with their action as transcription regulators of protein-coding genes (32). There is also the probability that lncRNAs act like a ‘sponge’-leading to miRNA recruitment into EVs, or possibly to provide the mechanisms required for microRNA loading (13).

These findings confirm both the diversity of lncRNAs and their participation in carcinogenesis by demonstrating their influence on the promotion of angiogenesis, protein stability or miRNA inhibition by acting as a sponge. In addition, as lncRNAs are detected in TEXs or apoptotic bodies, and have high stability during circulation, they could be considered as potential cancer biomarkers (33); however, some studies indicate they may display tumour suppression behavior through the blocking of the transcription of apoptosis-inhibiting genes and stimulation of p53 expression (34) or the structural modification of chromatin (35).

lncRNAs as protumorigenic molecules

Numerous studies implicate lncRNAs as cancer-associated molecules that are enriched in exosomes. A classic feature of the tumour microenvironment is hypoxia, which has a notable impact on cancer progression. Activation of its pathway via the transcription factor hypoxia inducible factor (HIF) plays a part in metastasis and a more aggressive phenotype. The role of lncRNAs in hypoxia-driven cancer progression came into view recently, where these hypoxia-responsive lncRNAs play a critical part in regulating hypoxic gene expressions (36). The next subsections will present the characteristics of the most frequent types.

HOTAIR: HOX Transcript Antisense RNA has been associated with progression and poor prognosis in patients with squamous cell carcinoma (SCC), hepatocellular carcinoma (HCC), urothelial bladder cancer (UBC) or colorectal cancer (CRC) (37). Yan et al (38) report the involvement of HOTAIR in the repression of WIF-1-a protein implicated in the Wnt signaling pathway. Moreover, epigenetic regulation is also achievable: It participates in the migration, invasion or proliferation, potentiating through silencing of miR-205 in bladder cancer. In addition, UBC cells displayed increased invasion with migration corresponding to regulation of the EMT pathway, which was confirmed by knockdown of HOTAIR. This resulted in reduced invasiveness by influencing genes connected with EMT such as MMP1 (Matrix metalloproteinase-1), ZEB1 (Zinc finger E-box-binding homeobox 1), TWIST1 (Twist-related protein 1), SNAI1 (Snail family zinc finger 1), ZO-1 (Zonula occludens 1), LAMC2 (Laminin, gamma 2), LAMB3 (Laminin, beta 3) or ABL2 (ABL proto-oncogene 2) (37). Studies by Xie et al, have shown that salivary HOTAIR could be used as biomarker in diagnostics: it had good discriminatory power for differentiating pancreatic cancer from healthy patients and benign pancreatic tumour (39, 40). In addition to its contribution in cancerous diseases, HOTAIR has also been described to play a role in rheumatoid arthritis, where its transcript activates MMP-2 and MMP-13 in synoviocytes and osteoclasts. This could promote joint and cartilage matrix cessation advancing joint destruction. Furthermore, HOTAIR-loaded exosomes with enriched lncRNA activate macrophages and influence immune response (41).

MALAT-1: Metastasis-Associated Lung Adenocarcinoma Transcript 1 or Nuclear-Enriched Abundant Transcript 2 (NEAT-2) was the first lncRNA to be associated with metastasis in Non-Small Cell Lung Cancer (NSCLC). This transcript, similar to the HOTAIR lncRNA, is also connected with migration or tumour growth induction. Zhang et al (42), indicated increased expression of transcript compared to healthy controls, and an association with higher TNM stage in patients with cancer. In vitro study knockdown of MALAT-1 in cancer cell lines significantly inhibited cell proliferation and colony formation, although it also induced cell cycle arrest and cell apoptosis due to a decrease in S phase progression. In addition, cyclin D1 and cyclin D2 downregulation was also observed, suggesting that MALAT-1 was involved in acceleration of the cell cycle. This indicates that MALAT-1 can be an important therapeutic target when developing treatments for NSCLC patients, because of its participation in the malignant phenotype of cancer through gene expression regulation (43). Alongside NSCLC, MALAT-1 has been proposed as a mediator of angiogenesis in Thyroid Cancer. Huang et al (44), found it to play a role in the secretion of the fibroblast growth factor 2 (FGF) protein from tumour-associated macrophages (TAMs), which led to vascularization, promotion of proliferation, invasion and migration of cancer cells. Nonetheless, MALAT-1 participation in releasing inflammatory cytokines or FGF2 secretion was predominantly blocked by overexpression of FGF2.

ZFAS1: Initial studies concerning the Zinc Finger AntiSense 1 (ZFAS1) lncRNA refer to alveolar development in the mammary gland, with a proposed role in the regulation of alveolar development and epithelial cell differentiation in the mammary gland (45). The transcript has been found to promote cancer progression by cell cycle regulation through cyclin-dependent kinase 1 (CDK1) interaction or destabilization of the p53 protein. The level of ZFAS1 was also upregulated in exosomes from HCC, CRC and gastric cancer (GC). Pan et al (46), noticed that overexpression of this transcript leads to proliferation and migration of GC cells and ZFAS1-positive exosomes which can be internalized by MKN-28 cells. Knockdown of ZFAS1 inhibited the migration, proliferation and EMT process; however, it also induced cell cycle arrest and programmed cell death, serving as a potential tumour suppressor. These findings have been attributed to decreased ERK, Bcl-2 and increased Bax protein levels. Additionally, ZFAS1 knockdown was observed to suppress cell migration by decreasing the expression of EMT transcription factors such as Slug or Twist. These findings suggest that ZFAS1 could promote metastasis by cell cycle acceleration or support EMT playing a pivotal role in GC diagnosis (47).

H19: H19 lncRNA is also involved in tumor metastasis and has been described as a master factor in cancer biology due to its frequent deregulation in almost all tumours, both in the initiation and progression steps (48). H19 is connected with the downregulation of E-Cadherin, which influences cell adhesion and leads to spread of cancer cells. In addition, Conigliaro et al (49) investigated H19 as a pro-angiogenic molecule promoting cell-to-cell adhesion (CD90+ cells with endothelial cell monolayer) and association with increased expression of VEGF and ICAM1 transcripts which in turn leads to new vessel development. H19 may also act as an epigenetic modulator or as miRNA sponge (50). In conclusion, this type of lncRNA could have a range of impacts on target cells and some of its functions still remain unknown; nevertheless, H19 has been proposed as a potential therapeutic target (e.g., in HCC) (51).

POU3F3: In vitro studies indicate that this transcript increases viability and cell proliferation in gliomas and its overexpression is associated with tumour grading (52). Hu et al (53) investigated whether exosomal POU3F3 from glioma cells could promote angiogenesis and its effect on endothelial cells. They indicate that lncRNA enriched the exosomes regulating the migration, proliferation and angiogenesis of Human Brain Microvascular Endothelial Cells (HBMECs) in both in vitro and in vivo studies. Silencing POU3F3 using shRNA decreased the migratory ability of endothelial cells but did not affect their motility. POU3F3 overexpression in HBMECs caused by lncRNA enriched exosome internalization, and significantly upregulated the transcript level of bFGF, bFGFR, Angio and VEGFA. These findings indicate that POU3F3 lncRNA can be an essential molecule in cell-to-cell communication or a regulator of angiogenesis in glioma cells (53).

TUC339: One of lncRNA transcribed from ultraconserved elements (UCEs) is TUC339. UCEs are 100% maintained genomic sequences that are greater than 200 bases in length and are conserved across human, mouse and rat genomes (54). The localization of these elements is connected with fragile sites and cancer-associated regions of the genome. Overexpression of this type of lncRNA has been reported in HCC (55). A study on the role of exosomal ultraconserved lncRNA in modulating target cell phenotype as a result of genomic changes found that TUC339 upregulation in HCC cell lines resulted in an enhancement of cell growth and a reduction of adhesion to cell-extracellular matrix (ECM). The knockdown of TUC339 decreased proliferation and altered the expression of a total of 843 genes, 469 of which were inhibited (56).

ROR: LncRNA ROR participates in liver cancer through disruption in TGF-β pathway. Although TGF-β has been related to cancer chemoresistance, the molecular actions involved in this event remain unknown. Takahashi et al (57), identified a possibly mechanism by which lncRNA may mediate chemoresistance dependent on TGF-β. They indicate that TGF-β reduced the sensitivity of HCC cells to sorafenib or doxorubicin, with a further reduction of sorafenib-dependent caspase 3/7 activity, leading to inhibition of the apoptosis process. Moreover, the release of extracellular vesicles and packaging of lncRNA within vesicles were also modulated by TGF-β. One of the selectively enriched exosomal lncRNAs thought to be a potential mediator of chemoresistance is RNA-ROR. Using a molecular pathway connected with p53 dependent signaling, lincRNA can contribute to tumor development by inhibiting apoptosis through the repression of p53. Additionally, knockdown of this lncRNA results in increased expression of Caspase 8 and GADD45B, leading to apoptosis induction and growth arrest. These data suggest that lincRNA ROR can be an important mediator in cell-to-cell communication and its contribution in chemoresistance has been confirmed in research. Inhibition of this transcript resulted in reversion of drug resistance, leading to cell death (58).

VLDLR: One of the long-intergenic non-coding RNA transcripts that support HCC development is called VLDLR (59). By showing that knockdown of this RNA can result in cell cycle arrest in G1/S phase, Takahashi et al (60), confirmed that it regulates HCC cell proliferation. Moreover, the quantity of lncRNA was increased in both HCC cells and extracellular vesicles following chemotherapeutic stress caused by sorafenib, doxorubicin and camptothecin, thus enhancing cell viability. Furthermore, the extracellular transfer of VLDLR to adjacent tumour cells could participate in chemoresistance through the modulation of ABCG2 gene expression.

The summary of the other lncRNA molecules that participate in tumour development is presented in Table VI.

Table VI.

Functions of chosen lncRNA molecules in numerous tumour types [based on (91–102)].

Table VI.

Functions of chosen lncRNA molecules in numerous tumour types [based on (91–102)].

lncRNAFunctionTumor type(Refs.)
ARSRResistance to sunitinib; overexpression leads to poor response of renal cancer patients; promoting c-MET expressionRenal cell carcinoma (RCC)(91)
MEG3Cell cycle arrest; decreases apoptosis by regulation of miR-21Cervical cancer (CC)(92,93)
RMRPActs as a miR-206 sponge and modulate the cell cycle by regulating the Cyclin D2 expressionGastric cancer (GC)(94)
NEAT1Promotes cancer progression through regulating CDK6 mediated by influence on miR-107Laryngeal squamous Cell cancer (LSCC)(95)
UCA1Enhances cell proliferation, migration, invasion, EMT processBladder cancer (BC)(96)
HULCInduces angiogenesis and modulates VEGF expression; upregulates sphingosine kinase 1 (SPHK1) by miR-107/E2F1 pathwayLiver cancer (LC)(97,98)
TUG1Promotes radioresistance and EMT transition; enhance proliferation and migrationBladder cancer (BC) Pancreatic cancer (PC)(99,100)
HOTTIPIncreases the chemoresistance by Wnt/β-catenin pathway activationOsteosarcoma(101)
CCAT2Upregulates VEGFA and TGFβ, promotes angiogenesis, decreases apoptosis by elevated Bcl-2 expression and inhibition of Bax and caspase-3Glioma cancer(102)
Suppressor function of lncRNA

In contrast to the lncRNAs with pro-cancerous properties, there is also another group of transcripts that could support normal cells rather than stimulating their conversion into a neoplastic phenotype (61).

GAS5: Several studies have implied that the Growth Arrest Specific 5 gene molecule has not been connected with cancer progression, as its expression has been observed to be reduced in various cancer types (62,63). Pickard et al (64), noticed that GAS5 participates in apoptosis regulation in prostate cancer cells. They indicated that the high expression of GAS5 level reduces tumour cell survival and hence induces pro-apoptotic activity. On the other hand, when GAS5 was downregulated, programmed cell death was diminished but growth arrest was not. In prostate carcinoma GAS5 has been found to bind to the receptor domain and inhibit transcriptional stimulation, leading to increased death of tumour cells (65).

PARTICL: Some studies also confirmed the role of lncRNAs in the methylation process in response to radiation. One of this lncRNA is PARTICL (promoter of MAT2A antisense radiation-induced circulating long non-coding RNA) a trans-acting mediator of DNA, and histone lysine methylation, a clever component for gene silencing (66). The findings indicate that in low dose irradiation, PARTICL can enhance EZH2 (enhancer of zeste homolog 2) expression, which in turn catalyzes the addition of methyl groups to histone H3 at lysine 27. Moreover, there was also an observed increase in the activity of the DNMT1 enzyme (DNA methyltransferase 1), global methylome enhancement methylome regulation e.g., CpG island methylation of WWOX gene. Finally, speculation about PARTICL as an epigenetic modifying platform targeting chromatin structure modulation through recruitment of PRC2 (Polycomb Repressive Complex 2) has been proposed; this has been proposed to act by the binding of lncRNA to a subunit of PRC2 called SUZ12 (Suppressor of Zeste 12). These data indicate that PARTICL interlinks regulators of epigenetic modifications that may affect the suppression of transcription (67).

CCND1: Another lncRNA found in exosomes (68) and connected with ionizing radiation-dependent transcription (69) is ncRNA CCND1 (cyclin D1). This single strand transcript binds to the TLS (translocated in liposarcoma) modulator, thus recruiting it to CCND1 promoter and inducing negative regulation of gene transcription. In addition, it also blocks the activity of CREB (cAMP response element binding) and p300 proteins.

EXO1-4: The four molecules that occur abundantly in exosomes in cervical cancer were defined as Exo1-4 (70). Those transcripts contain DNase hypersensitive regions as well as RNAPII and CTCF binding sites (71). Exo1-4 is a subset of lncRNAs that modulate cell function through increasing recipient cell viability by binding to various proteins: For example, Exo2 directly interacts with lactate dehydrogenase B (LDHB) while Exo4 reacts with high mobility group protein 17 (HMG-17). This speculates that lncRNA Exo1-4 has an impact on processes like cell metabolism or nucleosomal architecture.

lncRNAs as novel class of tumour biomarkers

Because lncRNA has greater tissue specificity than mRNA (72), these molecules may act as specific diagnostic biomarkers (73). When included in extracellular vesicles, circulation of RNA in body fluids is stable and able to resist the activity of ribonucleases (74), which makes them potential cancer prognostic markers (75). The profiles of lncRNA varies depending on tumour types that may be useful for clinical research, and moreover, these molecules could replace biopsies owing to the reduced level of invasiveness (76). Exosomal lncRNA can be nominated as cancer biomarkers but some of them are responsible for the development of other diseases (77). A set of differently expressed lncRNA associated with non-cancerous or cancerous diseases is shown in Table VII.

Table VII.

Participation of lncRNAs in various diseases [based on (77, 103–110)].

Table VII.

Participation of lncRNAs in various diseases [based on (77, 103–110)].

lncRNADiseaseFunction(Refs.)
CRNDE-hColorectal cancer (CRC)Promotes metabolic changes through insulin/IGF signaling(103)
p21Prostate cancer (PCa)Alter gene expression by modulating mRNA translation and suppressing the p53 or Wnt/β-catenin pathway(104)
PCA3Prostate cancer (PCa)Modulates the survival of cells downstream of androgen receptor signaling(105)
BCAR4Breast cancer (BC)Convert phenotype into an estrogen-independent, antiestrogen-resistant; tamoxifen resistance via HER2 signaling(106)
ATBHepatocellular carcinoma (HCC)Suppresses E-cadherin, leading to progression of epithelial tumor cells via inducing EMT(107)
ANRILNon-small cell lung carcinoma (NSCLC)Affects proliferation and apoptosis using suppression of KLF2 and P21 transcription(108)
LINC00152Gastric cancer (GCA)Connects with depth of invasion among gastric cancer patients(109)
RP11-445H22.4Breast cancer (BC)Its expression levels correlated with estrogen receptor (ER), progesterone receptor (PR), and menopausal status of the breast cancer patients; this molecule showed a remarkable improvement compared with CEA(110)
RP11-1382.1Chronic kidney disease (CKD)Unknown function(77)

Potential usefulness and perspectives

As a result of maintaining full stability of lncRNA in extracellular vesicles as mentioned earlier, these transcripts can be taken up by target cells with their full functionality preserved (78). This make EVs effective nanocarriers that are capable of regulating lncRNAs expression in cancer cells (79). An attractive potential tumour therapy that requires further development maintaining high levels of tumour suppressor lncRNAs or destabilizing oncogenic ones (80). H19 is an example of the use of pro-tumorigenic long non-coding transcripts in the specific targeting of tumour cells. The regulatory sequence of this lncRNA was used in the successful formation of the BC-819 plasmid. In vivo this construct encodes the toxin Diphtheria Toxin A (DTA) which provided encouraging results in the treatment of bladder or colon cancer, as well as pancreas carcinoma or NSCLC (81). There are also potential approaches to inducing overexpression of lncRNA MEG3 (Maternally Expressed Gene 3) in lung adenocarcinoma in order to increase sensitivity to cisplatin-based chemotherapy (82). Another potential type of lncRNA-based therapy exploits the ‘sponge’ activity of those transcripts, resulting in lowered miRNA activity in tumours (81).

Looking prospectively, there are still many challenges to be tackled. Even following the identification of numerous lncRNAs connected with various diseases, there is still the need to identify more transcripts and clarify their mechanism of action. Unbiased genetic screening, lncRNA expression patterns and characterization of associated proteins should provide guidance. Moreover, the development of better technologies directed to improve lncRNA-based therapies will effectively expand their usage as therapeutic or diagnostic strategies. Finally, investigation of new tools using the epigenetic functionality of lncRNAs could help in the research and treatment of diseases that depend on epigenetics (83). When it comes to the development of exosomal lncRNA-based therapeutics, a few problems were identified including uncertain functionality in biological pathways, the pharmacokinetics and toxicity of lncRNAs and the precise quantification of EV-bounded non-coding transcripts which need to be addressed accordingly (84).

In conclusion, progressive studies of lncRNA properties can lead to their use as cancer biomarkers with high sensitivity and accuracy, resulting in early-stage detection, and improved performance of existing clinical biomarkers. Long non-coding RNA can either have a suppressive function in terms of cancer development, or promote its progression. A good example of transporters that carry lncRNAs are exosomes, participating in cell-to-cell communication and thereafter affecting physiological and pathological changes. There are still several challenges that needs to be tackled in order to provide efficient EV-based lncRNAs therapeutics for clinical application, but the current state of research offers promise.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

DK drafted the manuscript, designed the figures, and researched the literature. DK and EP designed the article. EP, AKB and RH revised the article. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Pan BT and Johnstone RM: Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell. 33:967–978. 1983. View Article : Google Scholar : PubMed/NCBI

2 

Johnstone RM, Adam M, Hammond JR, Orr L and Turbide C: Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Boil Chem. 262:9412–9420. 1987.

3 

Yu S, Cao H, Shen B and Feng J: Tumor-derived exosomes in cancer progression and treatment failure. Oncotarget. 6:37151–37168. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Isola AL and Chen S: Exosomes: The link between GPCR activation and metastatic potential? Front Genet. 7:562016. View Article : Google Scholar : PubMed/NCBI

5 

Ciardiello C, Cavallini L, Spinelli C, Yang J, Reis-Sobreiro M, de Candia P, Minciacchi VR and Di Vizio D: Focus on extracellular vesicles: New frontiers of cell-to-cell communication in cancer. Int J Mol Sci. 17:1752016. View Article : Google Scholar : PubMed/NCBI

6 

Taverna S, Giallombardo M, Gil-Bazo I, Carreca AP, Castiglia M, Chacartegui J, Araujo A, Alessandro R, Pauwels P, Peeters M and Rolfo C: Exosomes isolation and characterization in serum is feasible in non-small cell lung cancer patients: Critical analysis of evidence and potential role in clinical practice. Oncotarget. 7:28748–28760. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Fu H, Yang H, Zhang X and Xu W: The emerging roles of exosomes in tumor-stroma interaction. J Cancer Res Clin Oncol. 142:1897–1907. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Soung YH, Nguyen T, Cao H, Lee J and Chung J: Emerging roles of exosomes in cancer invasion and metastasis. BMB Rep. 49:18–25. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Rahman MA, Barger JF, Lovat F, Gao M, Otterson GA and Nana-Sinkam P: Lung cancer exosomes as drivers of epithelial mesenchymal transition. Oncotarget. 7:54852–54866. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Wang Y, Yi J, Chen X, Zhang Y, Xu M and Yang Z: The regulation of cancer cell migration by lung cancer cell-derived exosomes through TGF-β and IL-10. Oncol Lett. 11:1527–1530. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Bobrie A and Thery C: Unraveling the physiological functions of exosome secretion by tumors. Oncoimmunology. 2:e225652013. View Article : Google Scholar : PubMed/NCBI

12 

De Toro J, Herschlik L, Waldner C and Mongini C: Emerging roles of exosomes in normal and pathological conditions: New insights for diagnosis and therapeutic applications. Front Immunol. 6:2032015. View Article : Google Scholar : PubMed/NCBI

13 

Kim KM, Abdelmohsen K, Mustapic M, Kapogiannis D and Gorospe M: RNA in extracellular vesicles. Wiley Interdiscip Rev RNA. 8:2017. View Article : Google Scholar

14 

Neviani P and Fabbri M: Exosomic microRNAs in the tumor microenvironment. Front Med (Lausanne). 2:472015.PubMed/NCBI

15 

Martin F, Roth DM, Jans DA, Pouton CW, Partridge LJ, Monk PN and Moseley GW: Tetraspanins in viral infections: A fundamental role in viral biology? J Virol. 79:10839–10851. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Baietti MF, Zhang Z, Mortier E, Melchior A, Degeest G, Geeraerts A, Ivarsson Y, Depoortere F, Coomans C, Vermeiren E, et al: Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 14:677–685. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Fares J, Kashyap R and Zimmermann P: Syntenin: Key player in cancer exosome biogenesis and uptake? Cell Adh Migr. 11:124–126. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Ghossoub R, Lembo F, Rubio A, Gaillard CB, Bouchet J, Vitale N, Slavík J, Machala M and Zimmermann P: Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2. Nat Commun. 5:34772014. View Article : Google Scholar : PubMed/NCBI

19 

Shim S: Role and regulation of ESCRT-III In multivesiculr body biogenesis. All Theses and Dissertations (ETDs). 3232009.

20 

Roxrud I, Stenmark H and Malerod L: ESCRT & Co. Biol Cell. 102:293–318. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Votteler J and Sundquist WI: Virus budding and the ESCRT pathway. Cell Host Microbe. 14:232–241. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Amano M, Nakayama M and Kaibuchi K: Rho-kinase/ROCK: A key regulator of the cytoskeleton and cell polarity. Cytoskeleton (Hoboken). 67:545–554. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Roucourt B, Meeussen S, Bao J, Zimmermann P and David G: Heparanase activates the syndecan-syntenin-ALIX exosome pathway. Cell Res. 25:412–428. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Roma-Rodrigues C, Fernandes AR and Baptista PV: Exosome in tumour microenvironment: Overview of the crosstalk between normal and cancer cells. Biomed Res Int. 2014:1794862014. View Article : Google Scholar : PubMed/NCBI

25 

Gezer U, Ozgur E, Cetinkaya M, Isin M and Dalay N: Long non-coding RNAs with low expression levels in cells are enriched in secreted exosomes. Cell Biol Int. 38:1076–1079. 2014.PubMed/NCBI

26 

Schageman J, Zeringer E, Li M, Barta T, Lea K, Gu J, Magdaleno S, Setterquist R and Vlassov AV: The complete exosome workflow solution: From isolation to characterization of RNA cargo. Biomed Res Int. 2013:2539572013. View Article : Google Scholar : PubMed/NCBI

27 

Batagov AO, Kuznetsov VA and Kurochkin IV: Identification of nucleotide patterns enriched in secreted RNAs as putative cis-acting elements targeting them to exosome nano-vesicles. BMC Genomics. 3 (Suppl 12):S182011. View Article : Google Scholar

28 

Okazaki Y, Furuno M, Kasukawa T, Adachi J, Bono H, Kondo S, Nikaido I, Osato N, Saito R, Suzuki H, et al: Analysis of the mouse transcriptome based on functional annotation of 60,770 full-length cDNAs. Nature. 420:563–573. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, Stadler PF, Hertel J, Hackermüller J, Hofacker IL, et al: RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 316:1484–1488. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Perkel JM: Visiting ‘noncodarnia’. Biotechniques. 54:301 303–304. 2013. View Article : Google Scholar

31 

Wang KC and Chang HY: Molecular mechanisms of long noncoding RNAs. Mol Cell. 43:904–914. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Sato-Kuwabara Y, Melo SA, Soares FA and Calin GA: The fusion of two worlds: Non-coding RNAs and extracellular vesicles-diagnostic and therapeutic implications (Review). Int J Oncol. 46:17–27. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Bolha L, Ravnik-Glavac M and Glavac D: Long noncoding RNAs as biomarkers in cancer. Dis Markers. 2017:72439682017. View Article : Google Scholar : PubMed/NCBI

34 

Hewson C and Morris KV: Form and function of exosome-associated long non-coding RNAs in cancer. Curr Top Microbiol Immunol. 394:41–56. 2016.PubMed/NCBI

35 

Zhao J, Ohsumi TK, Kung JT, Ogawa Y, Grau DJ, Sarma K, Song JJ, Kingston RE, Borowsky M and Lee JT: Genome-wide identification of polycomb-associated RNAs by RIP-seq. Mol Cell. 40:939–953. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Shih JW and Kung HJ: Long non-coding RNA and tumor hypoxia: New players ushered toward an old arena. J Biomed Sci. 24:532017. View Article : Google Scholar : PubMed/NCBI

37 

Berrondo C, Flax J, Kucherov V, Siebert A, Osinski T, Rosenberg A, Fucile C, Richheimer S and Beckham CJ: Expression of the long non-coding RNA HOTAIR correlates with disease progression in bladder cancer and is contained in bladder cancer patient urinary exosomes. PLoS One. 11:e01472362016. View Article : Google Scholar : PubMed/NCBI

38 

Yan TH, Lu SW, Huang YQ, Que GB, Chen JH, Chen YP, et al: Upregulation of the long noncoding RNA HOTAIR predicts recurrence in stage Ta/T1 bladder cancer. Tumour biology: the journal of the International Society for Oncodevelopmental Biology and Medicine. 2014.35(10): 10249–57. View Article : Google Scholar : PubMed/NCBI

39 

Xie Z, Chen X, Li J, Guo Y, Li H, Pan X, Jiang J, Liu H and Wu B: Salivary HOTAIR and PVT1 as novel biomarkers for early pancreatic cancer. Oncotarget. 7:25408–25419. 2016.PubMed/NCBI

40 

Wang J, Zhang X, Ji C, Zhang L, Di Y, Lou W, Zhang X and X J: Novel implications of exosomes and lncRNAs in the diagnosis and treatment of pancreatic cancer. Novel Implications of Exosomes in Diagnosis and Treatment of Cancer and Infectious Diseases. Wang J; IntechOpen, : 2017, https://www.intechopen.com/books/novel-implications-of-exosomes-in-diagnosis-and-treatment-of-cancer-and-infectious-diseases/novel-implications-of-exosomes-and-lncrnas-in-the-diagnosis-and-treatment-of-pancreatic-cancer09th–May. 2019 View Article : Google Scholar

41 

Song J, Kim D, Han J, Kim Y, Lee M and Jin EJ: PBMC and exosome-derived Hotair is a critical regulator and potent marker for rheumatoid arthritis. Clin Exp Med. 15:121–126. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Zhang R, Xia Y, Wang Z, Zheng J, Chen Y, Li X, Wang Y and Ming H: Serum long non coding RNA MALAT-1 protected by exosomes is up-regulated and promotes cell proliferation and migration in non-small cell lung cancer. Biochem Biophys Res Commun. 490:406–414. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Gutschner T, Hammerle M, Eissmann M, Hsu J, Kim Y, Hung G, Revenko A, Arun G, Stentrup M, Gross M, et al: The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res. 73:1180–1189. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Huang JK, Ma L, Song WH, Lu BY, Huang YB, Dong HM, Ma XK, Zhu ZZ and Zhou R: LncRNA-MALAT1 promotes angiogenesis of thyroid cancer by modulating tumor-associated macrophage FGF2 protein secretion. J Cell Biochem. 118:4821–4830. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Askarian-Amiri ME, Crawford J, French JD, Smart CE, Smith MA, Clark MB, Ru K, Mercer TR, Thompson ER, Lakhani SR, et al: SNORD-host RNA Zfas1 is a regulator of mammary development and a potential marker for breast cancer. RNA. 17:878–891. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Pan L, Liang W, Fu M, Huang ZH, Li X, Zhang W, Zhang P, Qian H, Jiang PC, Xu WR and Zhang X: Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. J Cancer Res Clin Oncol. 143:991–1004. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Zhou H, Wang F, Chen H, Tan Q, Qiu S, Chen S, Jing W, Yu M, Liang C, Ye S and Tu J: Increased expression of long-noncoding RNA ZFAS1 is associated with epithelial-mesenchymal transition of gastric cancer. Aging (Albany NY). 8:2023–2038. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Matouk IJ, Halle D, Raveh E, Gilon M, Sorin V and Hochberg A: The role of the oncofetal H19 lncRNA in tumor metastasis: Orchestrating the EMT-MET decision. Oncotarget. 7:3748–3765. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Conigliaro A, Costa V, Lo Dico A, Saieva L, Buccheri S, Dieli F, Manno M, Raccosta S, Mancone C, Tripodi M, et al: CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol Cancer. 14:1552015. View Article : Google Scholar : PubMed/NCBI

50 

Kallen AN, Zhou XB, Xu J, Qiao C, Ma J, Yan L, Lu L, Liu C, Yi JS, Zhang H, et al: The imprinted H19 lncRNA antagonizes let-7 microRNAs. Mol Cell. 52:101–112. 2013. View Article : Google Scholar : PubMed/NCBI

51 

George J and Patel T: Noncoding RNA as therapeutic targets for hepatocellular carcinoma. Semin Liver Dis. 35:63–74. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Lang HL, Hu GW, Chen Y, Liu Y, Tu W, Lu YM, Wu L and Xu GH: Glioma cells promote angiogenesis through the release of exosomes containing long non-coding RNA POU3F3. Eur Rev Med Pharmacol Sci. 21:959–972. 2017.PubMed/NCBI

53 

Hu C, Chen M, Jiang R, Guo Y, Wu M and Zhang X: Exosome-related tumor microenvironment. Journal of Cancer. 2018.9(17): 3084–92. View Article : Google Scholar : PubMed/NCBI

54 

Mitra SA, Mitra AP and Triche TJ: A central role for long non-coding RNA in cancer. Front Genet. 3:172012. View Article : Google Scholar : PubMed/NCBI

55 

Braconi C, Valeri N, Kogure T, Gasparini P, Huang N, Nuovo GJ, Terracciano L, Croce CM and Patel T: Expression and functional role of a transcribed noncoding RNA with an ultraconserved element in hepatocellular carcinoma. Proc Natl Acad Sci USA. 108:786–791. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Kogure T, Yan IK, Lin WL and Patel T: Extracellular vesicle-mediated transfer of a novel long noncoding RNA TUC339: A mechanism of intercellular signaling in human hepatocellular cancer. Genes Cancer. 4:261–272. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Takahashi K, Yan IK, Kogure T, Haga H and Patel T: Extracellular vesicle-mediated transfer of long non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio. 4:458–467. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Li Y, Jiang B, Zhu H, Qu X, Zhao L, Tan Y, Jiang Y, Liao M and Wu X: Inhibition of long non-coding RNA ROR reverses resistance to Tamoxifen by inducing autophagy in breast cancer. Tumour Biol. 39:10104283177057902017. View Article : Google Scholar : PubMed/NCBI

59 

Yang N, Li S, Li G, Zhang S, Tang X, Ni S, Jian X, Xu C, Zhu J and Lu M: The role of extracellular vesicles in mediating progression, metastasis and potential treatment of hepatocellular carcinoma. Oncotarget. 8:3683–3695. 2017.PubMed/NCBI

60 

Takahashi K, Yan IK, Wood J, Haga H and Patel T: Involvement of extracellular vesicle long noncoding RNA (linc-VLDLR) in tumor cell responses to chemotherapy. Mol Cancer Res. 12:1377–1387. 2014. View Article : Google Scholar : PubMed/NCBI

61 

Fritah S, Niclou SP and Azuaje F: Databases for lncRNAs: A comparative evaluation of emerging tools. RNA. 20:1655–1665. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F and Williams GT: GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene. 28:195–208. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Lucafò M, Stocco G and Decorti GJTCR: Emerging molecular mechanisms underlying cancer metastasis: The rising role of the long non-coding RNA GAS5. Transl Cancer Res. 5 (Suppl 4):S827–S830. 2016. View Article : Google Scholar

64 

Pickard MR, Mourtada-Maarabouni M and Williams GT: Long non-coding RNA GAS5 regulates apoptosis in prostate cancer cell lines. Biochim Biophys Acta. 1832:1613–1623. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Kino T, Hurt DE, Ichijo T, Nader N and Chrousos GP: Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci Signal. 3:ra82010. View Article : Google Scholar : PubMed/NCBI

66 

O'Leary VB, Hain S, Maugg D, Smida J, Azimzadeh O, Tapio S, Ovsepian SV and Atkinson MJ: Long non-coding RNA PARTICLE bridges histone and DNA methylation. Sci Rep. 7:17902017. View Article : Google Scholar : PubMed/NCBI

67 

O'Leary VB, Ovsepian SV, Carrascosa LG, Buske FA, Radulovic V, Niyazi M, Moertl S, Trau M, Atkinson MJ and Anastasov N: PARTICLE, a triplex-forming long ncrna, regulates locus-specific methylation in response to low-dose irradiation. Cell Rep. 11:474–485. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Deng J, Tang J, Wang G and Zhu YS: Long non-coding RNA as potential biomarker for prostate cancer: Is it making a difference? Int J Environ Res Public Health. 14:E2702017. View Article : Google Scholar : PubMed/NCBI

69 

Wang X, Arai S, Song X, Reichart D, Du K, Pascual G, Tempst P, Rosenfeld MG, Glass CK and Kurokawa R: Induced ncRNAs allosterically modify RNA-binding proteins in cis to inhibit transcription. Nature. 454:126–130. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Di Liegro CM, Schiera G and Di Liegro I: Extracellular vesicle-associated RNA as a carrier of epigenetic information. Genes (Basel). 8:E2402017. View Article : Google Scholar : PubMed/NCBI

71 

Hewson C, Capraro D, Burdach J, Whitaker N and Morris KV: Extracellular vesicle associated long non-coding RNAs functionally enhance cell viability. Noncoding RNA Res. 1:3–11. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, et al: The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 22:1775–1789. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Yarmishyn AA and Kurochkin IV: Long noncoding RNAs: A potential novel class of cancer biomarkers. Front Genet. 6:1452015. View Article : Google Scholar : PubMed/NCBI

74 

Shi T, Gao G and Cao Y: Long noncoding RNAs as novel biomarkers have a promising future in cancer diagnostics. Dis Markers. 2016:90851952016. View Article : Google Scholar : PubMed/NCBI

75 

Akers JC, Gonda D, Kim R, Carter BS and Chen CC: Biogenesis of extracellular vesicles (EV): Exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J Neurooncol. 113:1–11. 2013. View Article : Google Scholar : PubMed/NCBI

76 

Silva A, Bullock M and Calin G: The clinical relevance of long non-coding RNAs in cancer. Cancers (Basel). 7:2169–2182. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Khurana R, Ranches G, Schafferer S, Lukasser M, Rudnicki M, Mayer G and Hüttenhofer A: Identification of urinary exosomal noncoding RNAs as novel biomarkers in chronic kidney disease. RNA. 23:142–152. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Takahashi K, Yan IK, Haga H and Patel T: Modulation of hypoxia-signaling pathways by extracellular linc-RoR. J Cell Sci. 127:1585–1594. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Gutschner T, Richtig G, Haemmerle M and Pichler M: From biomarkers to therapeutic targets-the promises and perils of long non-coding RNAs in cancer. Cancer Metastasis Rev. 37:83–105. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Rao AKDM, Rajkumar T and Mani S: Perspectives of long non-coding RNAs in cancer. Mol Boil Rep. 44:203–18. 2017. View Article : Google Scholar

81 

Smaldone MC and Davies BJ: BC-819, a plasmid comprising the H19 gene regulatory sequences and diphtheria toxin A, for the potential targeted therapy of cancers. Curr Opin Mol Ther. 12:607–616. 2010.PubMed/NCBI

82 

Liu J, Wan L, Lu K, Sun M, Pan X, Zhang P, Lu B, Liu G and Wang Z: The long noncoding RNA MEG3 contributes to cisplatin resistance of human lung adenocarcinoma. PLoS One. 10:e01145862015. View Article : Google Scholar : PubMed/NCBI

83 

Wu R, Su Y, Wu H, Dai Y, Zhao M and Lu Q: Characters, functions and clinical perspectives of long non-coding RNAs. Mol Genet Genomics. 291:1013–1033. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Qi P, Zhou XY and Du X: Circulating long non-coding RNAs in cancer: Current status and future perspectives. Mol Cancer. 15:392016. View Article : Google Scholar : PubMed/NCBI

85 

Schmidt O and Teis D: The ESCRT machinery. Curr Biol. 22:R116–R120. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Keller S, Sanderson MP, Stoeck A and Altevogt P: Exosomes: From biogenesis and secretion to biological function. Immunol Lett. 107:102–108. 2006. View Article : Google Scholar : PubMed/NCBI

87 

Ruiz-Martinez M, Navarro A, Marrades RM, Vinolas N, Santasusagna S, Munoz C, Ramírez J, Molins L and Monzo M: YKT6 expression, exosome release, and survival in non-small cell lung cancer. Oncotarget. 7:51515–51524. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, et al: Tumour exosome integrins determine organotropic metastasis. Nature. 527:329–335. 2015. View Article : Google Scholar : PubMed/NCBI

89 

Yang C and Robbins PD: The roles of tumor-derived exosomes in cancer pathogenesis. Clin Dev Immunol. 2011:8428492011. View Article : Google Scholar : PubMed/NCBI

90 

Zhang A, Zhou N, Huang J, Liu Q, Fukuda K, Ma D, Lu Z, Bai C, Watabe K and Mo YY: The human long non-coding RNA-RoR is a p53 repressor in response to DNA damage. Cell Res. 23:340–350. 2013. View Article : Google Scholar : PubMed/NCBI

91 

Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, Chen W, Liu F, Sun W, Li XF, et al: Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell. 29:653–668. 2016. View Article : Google Scholar : PubMed/NCBI

92 

Zhang J, Liu SC, Luo XH, Tao GX, Guan M, Yuan H and Hu DK: Exosomal long noncoding RNAs are differentially expressed in the cervicovaginal lavage samples of cervical cancer patients. J Clin Lab Anal. 30:1116–1121. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Zhang J, Yao T, Wang Y, Yu J, Liu Y and Lin Z: Long noncoding RNA MEG3 is downregulated in cervical cancer and affects cell proliferation and apoptosis by regulating miR-21. Cancer Boil Ther. 17:104–113. 2016. View Article : Google Scholar

94 

Shao Y, Ye M, Li Q, Sun W, Ye G, Zhang X, Yang Y, Xiao B and Guo J: LncRNA-RMRP promotes carcinogenesis by acting as a miR-206 sponge and is used as a novel biomarker for gastric cancer. Oncotarget. 7:37812–37824. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Wang P, Wu T, Zhou H, Jin Q, He G, Yu H, Xuan L, Wang X, Tian L, Sun Y, et al: Long noncoding RNA NEAT1 promotes laryngeal squamous cell cancer through regulating miR-107/CDK6 pathway. J Exp Clin Cancer Res. 35:222016. View Article : Google Scholar : PubMed/NCBI

96 

Xue M, Chen W, Xiang A, Wang R, Chen H, Pan J, Pang H, An H, Wang X, Hou H and Li X: Hypoxic exosomes facilitate bladder tumor growth and development through transferring long non-coding RNA-UCA1. Mol Cancer. 16:1432017. View Article : Google Scholar : PubMed/NCBI

97 

Juni RP, Abreu RC and da Costa Martins PA: Regulation of microvascularization in heart failure-an endothelial cell, non-coding RNAs and exosome liaison. Noncoding RNA Res. 2:45–55. 2017. View Article : Google Scholar : PubMed/NCBI

98 

Lu Z, Xiao Z, Liu F, Cui M, Li W, Yang Z, Li J, Ye L and Zhang X: Long non-coding RNA HULC promotes tumor angiogenesis in liver cancer by up-regulating sphingosine kinase 1 (SPHK1). Oncotarget. 7:241–254. 2016.PubMed/NCBI

99 

Tan J, Qiu K, Li M and Liang Y: Double-negative feedback loop between long non-coding RNA TUG1 and miR-145 promotes epithelial to mesenchymal transition and radioresistance in human bladder cancer cells. FEBS Lett. 589:3175–3181. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Qin CF and Zhao FL: Long non-coding RNA TUG1 can promote proliferation and migration of pancreatic cancer via EMT pathway. Eur Rev Med Pharmacol Sci. 21:2377–2384. 2017.PubMed/NCBI

101 

Li Z, Zhao L and Wang Q: Overexpression of long non-coding RNA HOTTIP increases chemoresistance of osteosarcoma cell by activating the Wnt/β-catenin pathway. Am J Transl Res. 8:2385–2393. 2016.PubMed/NCBI

102 

Lang HL, Hu GW, Zhang B, Kuang W, Chen Y, Wu L and Xu GH: Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2. Oncol Rep. 38:785–798. 2017. View Article : Google Scholar : PubMed/NCBI

103 

Liu T, Zhang X, Gao S, Jing F, Yang Y, Du L, Zheng G, Li P, Li C and Wang C: Exosomal long noncoding RNA CRNDE-h as a novel serum-based biomarker for diagnosis and prognosis of colorectal cancer. Oncotarget. 7:85551–85563. 2016. View Article : Google Scholar : PubMed/NCBI

104 

Isin M, Uysaler E, Ozgur E, Koseoglu H, Sanli O, Yucel OB, Gezer U and Dalay N: Exosomal lncRNA-p21 levels may help to distinguish prostate cancer from benign disease. Front Genet. 6:1682015.PubMed/NCBI

105 

Wang Y, Liu XJ and Yao XD: Function of PCA3 in prostate tissue and clinical research progress on developing a PCA3 score. Chin J Cancer Res. 26:493–500. 2014.PubMed/NCBI

106 

Godinho M, Meijer D, Setyono-Han B, Dorssers LC and van Agthoven T: Characterization of BCAR4, a novel oncogene causing endocrine resistance in human breast cancer cells. J Cell Physiol. 226:1741–1749. 2011. View Article : Google Scholar : PubMed/NCBI

107 

Jang SY, Kim G, Park SY, Lee YR, Kwon SH, Kim HS, Yoon JS, Lee JS, Kweon YO, Ha HT, et al: Clinical significance of lncRNA-ATB expression in human hepatocellular carcinoma. Oncotarget. 8:78588–78597. 2017. View Article : Google Scholar : PubMed/NCBI

108 

Nie FQ, Sun M, Yang JS, Xie M, Xu TP, Xia R, Liu YW, Liu XH, Zhang EB, Lu KH and Shu YQ: Long noncoding RNA ANRIL promotes non-small cell lung cancer cell proliferation and inhibits apoptosis by silencing KLF2 and P21 expression. Mol Cancer Ther. 14:268–277. 2015. View Article : Google Scholar : PubMed/NCBI

109 

Pang Q, Ge J, Shao Y, Sun W, Song H, Xia T, Xiao B and Guo J: Increased expression of long intergenic non-coding RNA LINC00152 in gastric cancer and its clinical significance. Tumour Biol. 35:5441–5447. 2014. View Article : Google Scholar : PubMed/NCBI

110 

Xu N, Chen F, Wang F, Lu X, Wang X, Lv M and Lu C: Clinical significance of high expression of circulating serum lncRNA RP11-445H22.4 in breast cancer patients: A Chinese population-based study. Tumour Biol. 36:7659–7665. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 20 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kołat D, Hammouz R, Bednarek AK and Płuciennik E: Exosomes as carriers transporting long non‑coding RNAs: Molecular characteristics and their function in cancer (Review). Mol Med Rep 20: 851-862, 2019
APA
Kołat, D., Hammouz, R., Bednarek, A.K., & Płuciennik, E. (2019). Exosomes as carriers transporting long non‑coding RNAs: Molecular characteristics and their function in cancer (Review). Molecular Medicine Reports, 20, 851-862. https://doi.org/10.3892/mmr.2019.10340
MLA
Kołat, D., Hammouz, R., Bednarek, A. K., Płuciennik, E."Exosomes as carriers transporting long non‑coding RNAs: Molecular characteristics and their function in cancer (Review)". Molecular Medicine Reports 20.2 (2019): 851-862.
Chicago
Kołat, D., Hammouz, R., Bednarek, A. K., Płuciennik, E."Exosomes as carriers transporting long non‑coding RNAs: Molecular characteristics and their function in cancer (Review)". Molecular Medicine Reports 20, no. 2 (2019): 851-862. https://doi.org/10.3892/mmr.2019.10340